Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Int J Mol Sci ; 25(13)2024 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-38999983

RESUMEN

The synthesis, biochemical evaluation and radiosynthesis of a cyclin-dependent kinases 4 and 6 (CDK4/6) inhibitor and radioligand was performed. NT431, a newly synthesized 4-fluorobenzyl-abemaciclib, exhibited high potency to CDK4/6 and against four cancer cell lines with IC50 similar to that of the parent abemaciclib. We performed a two-step one-pot radiosynthesis to produce [18F]NT431 with good radiochemical yield (9.6 ± 3%, n = 3, decay uncorrected), high radiochemical purity (>95%), and high molar activity (>370 GBq/µmol (>10.0 Ci/µmol). In vitro autoradiography confirmed the specific binding of [18F]NT431 to CDK4/6 in brain tissues. Dynamic PET imaging supports that both [18F]NT431 and the parent abemaciclib crossed the BBB albeit with modest brain uptake. Therefore, we conclude that it is unlikely that NT431 or abemaciclib (FDA approved drug) can accumulate in the brain in sufficient concentrations to be potentially effective against breast cancer brain metastases or brain cancers. However, despite the modest BBB penetration, [18F]NT431 represents an important step towards the development and evaluation of a new generation of CDK4/6 inhibitors with superior BBB penetration for the treatment and visualization of CDK4/6 positive tumors in the CNS. Also, [18F]NT431 may have potential application in peripheral tumors such as breast cancer and other CDK4/6 positive tumors.


Asunto(s)
Aminopiridinas , Bencimidazoles , Neoplasias Encefálicas , Quinasa 4 Dependiente de la Ciclina , Quinasa 6 Dependiente de la Ciclina , Tomografía de Emisión de Positrones , Inhibidores de Proteínas Quinasas , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/metabolismo , Humanos , Tomografía de Emisión de Positrones/métodos , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/enzimología , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/metabolismo , Línea Celular Tumoral , Bencimidazoles/farmacología , Bencimidazoles/química , Aminopiridinas/química , Aminopiridinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/síntesis química , Animales , Radiofármacos/química , Radioisótopos de Flúor/química , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Ratones , Femenino
2.
Ann Surg Oncol ; 30(2): 804-813, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36344711

RESUMEN

BACKGROUND: Nanoliposomal irinotecan (nal-IRI) is a promising novel hyperthermic intraperitoneal chemotherapy (HIPEC) agent given its enhanced efficacy against gastrointestinal tumors, safety profile, thermo-synergy, and heat stability. This report describes the first in-human phase 1 clinical trial of nal-IRI during cytoreductive surgery (CRS) and HIPEC. METHODS: Patients with peritoneal surface disease (PSD) from appendiceal and colorectal neoplasms were enrolled in a 3 + 3 dose-escalation trial using nal-IRI (70-280 mg/m2) during HIPEC for 30 min at 41 ± 1 °C. The primary outcome was safety. The secondary outcomes were pharmacokinetics (PK) and disease-free survival. Adverse events (AEs) categorized as grade 2 or higher were recorded. The serious AEs (SAEs) were mortality, grade ≥ 3 AEs, and dose-limiting toxicity (DLT). Irinotecan and active metabolite SN38 were measured in plasma and peritoneal washings. RESULTS: The study enrolled 18 patients, who received nal-IRI during HIPEC at 70 mg/m2 (n = 3), 140 mg/m2 (n = 6), 210 mg/m2 (n = 3), and 280 mg/m2 (n = 6). No DLT or mortality occurred. The overall morbidity for CRS/HIPEC was 39% (n = 7). Although one patient experienced neutropenia, no AE (n = 131) or SAE (n = 3) was definitively attributable to nal-IRI. At 280 mg/m2, plasma irinotecan and SN38 measurements showed maximum concentrations of 0.4 ± 0.6 µg/mL and 3.0 ± 2.4 ng/mL, a median time to maximum concentration of 24.5 and 26 h, and areas under the curve of 22.6 h*µg/mL and 168 h*ng/mL, respectively. At the 6-month follow-up visit, 83% (n = 15) of the patients remained disease-free. CONCLUSIONS: In this phase 1 HIPEC trial (NCT04088786), nal-IRI was observed to be safe, and PK profiling showed low systemic absorption overall. These data support future studies testing the efficacy of nal-IRI in CRS/HIPEC.


Asunto(s)
Neoplasias Colorrectales , Hipertermia Inducida , Neoplasias Peritoneales , Humanos , Irinotecán/uso terapéutico , Terapia Combinada , Calor , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Procedimientos Quirúrgicos de Citorreducción/efectos adversos , Neoplasias Colorrectales/patología , Hipertermia Inducida/efectos adversos , Tasa de Supervivencia
3.
Br J Cancer ; 123(3): 495, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32393850

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

4.
Br J Cancer ; 120(1): 88-96, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30377341

RESUMEN

BACKGROUND: Pancreatic cancer (PC) hijacks innate cellular processes to promote cancer growth. We hypothesized that PC exploits PD-1/PD-L1 not only to avoid immune responses, but to directly enhance growth. We also hypothesized that immune checkpoint inhibitors (ICIs) have direct cytotoxicity in PC. We sought to elucidate therapeutic targeting of PD-1/PD-L1. METHODS: PD-1 was assessed in PC cells, patient-derived organoids (PDOs), and clinical tissues. Then, PC cells were exposed to PD-L1 to evaluate proliferation. To test PD-1/PD-L1 signaling, cells were exposed to PD-L1 and MAPK was examined. Radio-immunoconjugates with anti-PD-1 drugs were developed to test uptake in patient-derived tumor xenografts (PDTXs). Next, PD-1 function was assessed by xenografting PD-1-knockdown cells. Finally, PC models were exposed to ICIs. RESULTS: PD-1 expression was demonstrated in PCs. PD-L1 exposure increased proliferation and activated MAPK. Imaging PDTXs revealed uptake of radio-immunoconjugates. PD-1 knockdown in vivo revealed 67% smaller volumes than controls. Finally, ICI treatment of both PDOs/PDTXs demonstrated cytotoxicity and anti-MEK1/2 combined with anti-PD-1 drugs produced highest cytotoxicity in PDOs/PDTXs. CONCLUSIONS: Our data reveal PCs innately express PD-1 and activate druggable oncogenic pathways supporting PDAC growth. Strategies directly targeting PC with novel ICI regimens may work with adaptive immune responses for optimal cytotoxicity.


Asunto(s)
Antígeno B7-H1/inmunología , Inmunoterapia , Neoplasias Pancreáticas/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/inmunología , Animales , Antígeno B7-H1/antagonistas & inhibidores , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Masculino , Ratones , Organoides/efectos de los fármacos , Organoides/inmunología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Cultivo Primario de Células , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Ann Surg Oncol ; 25(9): 2767-2775, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30003451

RESUMEN

BACKGROUND: Organoids are three-dimensional in vitro models of human disease developed from benign and malignant gastrointestinal tissues with tremendous potential for personalized medicine applications. We sought to determine whether gastric cancer patient-derived organoids (PDOs) could be safely established from endoscopic biopsies for rapid drug screening. METHODS: Patients underwent esophagogastroduodenoscopy (EGD) for surveillance or staging and had additional forceps biopsies taken for PDO creation. Cancer tissues from operative specimens were also used to create PDOs. To address potential tumor heterogeneity, we performed low-coverage whole-genome sequencing of endoscopic-derived PDOs with paired surgical PDOs and whole-tumor lysates. The stability of genomic alterations in endoscopic organoids was assessed by next-generation sequencing and nested polymerase chain reaction (PCR) assay. The feasibility and potential accuracy of drug sensitivity screening with endoscopic-derived PDOs were also evaluated. RESULTS: Gastric cancer PDOs (n = 15) were successfully established from EGD forceps biopsies (n = 8) and surgical tissues (n = 7) from five patients with gastric adenocarcinoma. Low-coverage whole-genomic profiling of paired EGD and surgical PDOs along with whole-tumor lysates demonstrated absence of tumor heterogeneity. Nested PCR assay identified similar KRAS alterations in primary tumor and paired organoids. Drug sensitivity testing of endoscopic-derived PDOs displayed standard dose-response curves to current gastric cancer cytotoxic therapies. CONCLUSIONS: Our study results demonstrate the feasibility of developing gastric cancer PDOs from EGD biopsies. These results also indicate that endoscopic-derived PDOs are accurate surrogates of the primary tumor and have the potential for drug sensitivity screening and personalized medicine applications.


Asunto(s)
Adenocarcinoma/patología , Antineoplásicos/farmacología , Endoscopía del Sistema Digestivo/métodos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Cultivo de Órganos/métodos , Organoides/patología , Neoplasias Gástricas/patología , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/cirugía , Biomarcadores de Tumor/genética , Biopsia , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Genómica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Organoides/efectos de los fármacos , Organoides/metabolismo , Medicina de Precisión , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/cirugía , Células Tumorales Cultivadas
7.
Sci Rep ; 14(1): 15100, 2024 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-38956204

RESUMEN

The design and radiosynthesis of [18F]NT376, a high potency inhibitor of class-IIa histone deacetylases (HDAC) is reported. We utilized a three-step radiochemical approach that led to the radiosynthesis of [18F]NT376 in a good radiochemical yield, (17.0 ± 3%, decay corrected), high radiochemical purity (> 97%) and relatively high molar activity of 185.0 GBq/µmol (> 5.0 Ci/µmol). The repositioning of the 18F-radiolabel into a phenyl ring (18F-Fluoro-aryl) of the class-IIa HDAC inhibitor avoided the shortcomings of the direct radiolabeling of the 5-trifluoromethyl-1,2,4-oxadiazole moiety that was reported by us previously and was associated with low molar activity (0.74-1.51 GBq/µmol, 20-41 mCi/µmol). This radiochemical approach could find a wider application for radiolabeling similar molecules with good radiochemical yield and high molar activity.


Asunto(s)
Radioisótopos de Flúor , Inhibidores de Histona Desacetilasas , Radiofármacos , Inhibidores de Histona Desacetilasas/química , Inhibidores de Histona Desacetilasas/síntesis química , Inhibidores de Histona Desacetilasas/farmacología , Radioisótopos de Flúor/química , Radiofármacos/química , Radiofármacos/síntesis química , Diseño de Fármacos , Humanos , Radioquímica/métodos , Oxadiazoles/química , Oxadiazoles/síntesis química
8.
Conn Med ; 77(4): 205-10, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23691733

RESUMEN

BACKGROUND: We previously demonstrated that pancreatic transection with a reinforced staple line results in significantly lower fistula rates than when stapling without reinforcement. (J Gastrointest Surg. 2007;11:345-349). Criticism of this initial study focused on the small size of the treated group (N = 13). We report four more years of experience with this technique with a larger sample size. METHODS: This was a before-after trial. Patients included had distal pancreatectomies with stapled stump closure. The main intervention analyzed was staple-line reinforcement with Seamguard. The experimental group consisted of a consecutive series of stapled pancreatectomies with reinforcement performed from 2005 to 2010. The control group was a consecutive series of stapled pancreatectomies without reinforcement performed between 2003 and 2005 (previously published). The main outcome measure was pancreatic fistula. RESULTS: 54 patients were included; 36 in the experimental group and 18 in the control group. Mean age was 62; 50% were males. The most common diagnoses were adenocarcinoma (31%), cystic neoplasm (24%), and neuroendocrine tumor (22%). There were no mortalities. Postoperative pancreatic leak rate was 39% in the control group, and 8% in the experimental group (P = 0.01). Seven of ten patients with leak required additional drain placement. Development of pancreatic leak resulted in prolonged hospital stays (12 vs eight days, P < 0.007). CONCLUSION: We demonstrate sustained success of reinforced stapling for pancreatic stump closure. Our technique is straightforward and results in reduced morbidity and cost. Our results suggest that surgical drains may not be needed when this technique is applied.


Asunto(s)
Pancreatectomía/métodos , Fístula Pancreática/epidemiología , Complicaciones Posoperatorias/epidemiología , Grapado Quirúrgico/métodos , Implantes Absorbibles , Adenocarcinoma/cirugía , Femenino , Humanos , Tiempo de Internación/estadística & datos numéricos , Masculino , Persona de Mediana Edad , Neoplasias Quísticas, Mucinosas y Serosas/cirugía , Tumores Neuroendocrinos/cirugía , Pancreatectomía/instrumentación , Neoplasias Pancreáticas/cirugía , Mallas Quirúrgicas , Resultado del Tratamiento
9.
Conn Med ; 76(4): 213-8, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22611720

RESUMEN

OBJECTIVE: Morbidity after pancreaticoduodenectomy (PD) is nearly 50%. In this study we analyzed if early enteral nutrition via feeding tube (FT) contributes to better patient outcomes. MATERIALS AND METHODS: Patients undergoing PD from 2003-2010. FTs were placed routinely before August 2006, and omitted thereafter. Short-term outcome measures included: time to start of oral diet, need for total parenteral nutrition (TPN), morbidity and mortality, pancreatic fistula, complications from FT, hospital length of stay, and disposition. Long-term outcome measures included time to start adjuvant therapy, and survival. RESULTS: N = 59 (25 had FT, 34 did not). Adenocarcinoma was found in 88%. Early institution of tube feeding had no positive impact on any of the outcome measures. There were three FT-related complications. CONCLUSIONS: Our results demonstrate that FT placement does not improve short-term or long-term outcomes after PD. Moreover, major complications can result from FT placement. We do not advocate the routine use of FT after PD.


Asunto(s)
Neoplasias Duodenales/cirugía , Nutrición Enteral , Intubación Gastrointestinal , Neoplasias Pancreáticas/cirugía , Pancreaticoduodenectomía , Adulto , Estudios de Cohortes , Femenino , Humanos , Masculino , Persona de Mediana Edad , Resultado del Tratamiento
10.
Blood ; 112(4): 1424-33, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18541724

RESUMEN

Epigenetic changes have been implicated in silencing several B-cell genes in Hodgkin and Reed-Sternberg cells (HRS) of Hodgkin lymphoma (HL), and this mechanism has been proposed to promote HRS survival and escape from immunosurveillance. However, the molecular and functional consequences of histone deacetylase (HDAC) inhibition in HL have not been previously described. In this study, we report that the HDAC inhibitor vorinostat induced p21 expression and decreased Bcl-xL levels causing cell-cycle arrest and apoptosis. Furthermore, vorinostat inhibited STAT6 phosphorylation and decreased its mRNA levels in a dose- and time-dependent manner, which was associated with a decrease in the expression and secretion of Thymus and Activation-Regulated Chemokine (TARC/CCL17) and interleukin (IL)-5 and an increase in IP-10 levels. Moreover, vorino-stat inhibited TARC secretion by dendritic cells that were activated by the thymic stromal lymphopoietin (TSLP). Collectively, these data suggest that pharmacologic HDAC inhibition in HL may induce favorable antitumor activity by a direct antiproliferative effect on HRS cells, and possibly by an immune mediated effect by altering cytokine and chemokines secretion in the microenvironment.


Asunto(s)
Quimiocina CCL17/biosíntesis , Citocinas/biosíntesis , Enfermedad de Hodgkin/tratamiento farmacológico , Ácidos Hidroxámicos/farmacología , Factor de Transcripción STAT6/antagonistas & inhibidores , Células Th2/efectos de los fármacos , Antineoplásicos/farmacología , Muerte Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Inhibidores de Histona Desacetilasas , Enfermedad de Hodgkin/patología , Humanos , Factor de Transcripción STAT6/metabolismo , Células Th2/metabolismo , Vorinostat
11.
Blood ; 112(3): 711-20, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18497318

RESUMEN

B-cell chronic lymphocytic leukemia (B-CLL) is a lymphoproliferative disorder characterized by the surface expression of CD20, CD5 antigens, as well as the receptor CD40. Activation of CD40 by its ligand (CD40L) induces proliferation and rescues the cells from spontaneous and chemotherapy-induced apoptosis. CD40 activation also induces secretion of cytokines, such as IL-6, IL-10, TNF-alpha, IL-8, and GM-CSF, which are involved in tumor cell survival, migration, and interaction with cells in the tumor microenvironment. Here we demonstrate that in primary B-CLL tumor cells, the novel antagonist anti-CD40 monoclonal antibody, HCD122, inhibits CD40L-induced activation of signaling pathways, proliferation and survival, and secretion of cytokines. Furthermore, HCD122 is also a potent mediator of antibody-dependent cellular cytotoxicity (ADCC), lysing B-CLL cells more efficiently than rituximab in vitro, despite a significantly higher number of cell surface CD20 binding sites compared with CD40. Unlike rituximab, however, HCD122 (formerly CHIR-12.12) does not internalize upon binding to the cells. Our data suggest that HCD122 may inhibit B-CLL growth by blocking CD40 signaling and by ADCC-mediated cell lysis.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Antígenos CD40/inmunología , Leucemia de Células B/inmunología , Adulto , Anciano , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales de Origen Murino , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Sitios de Unión , Femenino , Humanos , Masculino , Persona de Mediana Edad , Rituximab , Transducción de Señal/efectos de los fármacos
12.
Exp Hematol ; 36(4): 443-50, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18343280

RESUMEN

OBJECTIVE: To investigate the mechanisms of antiproliferative effect induced by the mammalian target of rapamycin (mTOR) inhibitor temsirolimus in mantle cell lymphoma (MCL). MATERIALS AND METHODS: The antiproliferative effect of temsirolimus on three well-defined MCL cell lines was examined by the MTS assay. Induction of cell-cycle arrest, autophagy, and apoptosis were determined by fluorescence-activated cell sorting analysis. The molecular mechanisms underlining these effects were determined by Western blot. Synergy between temsirolimus and vorinostat were examined by MTS assay and the combination index was calculated. RESULTS: Temsirolimus has antiproliferative activity in three MCL cell lines in a dose- and time-dependent manner. Mechanistically, temsirolimus inhibited mTOR, as evidenced by inhibition of ribosomal S6 phosphorylation, and induced cell-cycle arrest in the G(0)/G(1) phase and a decrease in p21 expression without altering p27 or cyclin D1 levels. Furthermore, temsirolimus increased the number of acidic vesicular organelles and the amount of microtubule-associated protein 1 light-chain 3 processing, which are characteristic of autophagy, without induction of apoptosis. These changes were not associated with alteration in phosphorylated extracellular signal-regulated kinase (ERK), beclin-1, Bax, or Bak levels. In contrast, treatment of these cell lines with the histone deacetylase inhibitor vorinostat decreased ERK phosphorylation, activated caspase 3, and induced apoptosis. Moreover, temsirolimus synergized with submaximal concentrations of vorinostat in all MCL cell lines. CONCLUSION: This is the first report of temsirolimus-induced autophagy in MCL, and of vorinostat inhibition of ERK phosphorylation in MCL. Collectively, these data suggest that the combination of temsirolimus and vorinostat have synergistic antiproliferative activity in MCL cells by distinctively targeting apoptosis and autophagy.


Asunto(s)
Autofagia/efectos de los fármacos , Ciclina D1/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Ácidos Hidroxámicos/farmacología , Linfoma de Células del Manto/metabolismo , Sirolimus/análogos & derivados , Antineoplásicos/farmacología , Línea Celular , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Fase G1/efectos de los fármacos , Humanos , Linfoma de Células del Manto/tratamiento farmacológico , Sirolimus/farmacología , Vorinostat
13.
Haematologica ; 92(2): 269-70, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17296586

RESUMEN

B-lymphocyte stimulator (BLyS) plays a critical role in the survival of B-lymphocytes. In 50 patients with Hodgkin's lymphoma BLyS levels were higher in newly diagnosed patients (median 2.0 ng/mL, range <0.3-56.0) and relapsed patients (8.7 ng/mL, range 1.5-71.5) than in 93 healthy donors (<0.3 ng/mL, range <0.3-0.5). High serum BLyS levels (> or =2.0 ng/mL) in newly diagnosed patients were associated with resistance to therapy (p=0.01) and shorter progression-free survival (log-rank p=0.029, 2-year rate 64% vs 100%). Serum BLyS levels may have prognostic significance in Hodgkin's lymphoma.


Asunto(s)
Factor Activador de Células B/sangre , Factor Activador de Células B/fisiología , Enfermedad de Hodgkin/sangre , Enfermedad de Hodgkin/diagnóstico , Adulto , Estudios de Casos y Controles , Supervivencia sin Enfermedad , Femenino , Enfermedad de Hodgkin/terapia , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Recurrencia , Resultado del Tratamiento
14.
Clin Cancer Res ; 12(2): 584-90, 2006 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-16428504

RESUMEN

PURPOSE: Heat shock protein 90 (HSP90) is a chaperone for several client proteins involved in transcriptional regulation, signal transduction, and cell cycle control. HSP90 is abundantly expressed by a variety of tumor types and has been recently targeted for cancer therapy. The objective of this study was to determine the role of HSP90 in promoting growth and survival of Hodgkin's lymphoma and to determine the molecular consequences of inhibiting HSP90 function by the small-molecule 17-allylamino-17-demethoxy-geldanamycin (17-AAG) in Hodgkin's lymphoma. EXPERIMENTAL DESIGN: HSP90 expression in Hodgkin's lymphoma cell lines was determined by Western blot and in primary lymph node sections from patients with Hodgkin's lymphoma by immunohistochemistry. Cell viability was determined by the 3-(4,5-dimethyl-thiazol-2yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assay. Apoptosis and cell cycle fractions were determined by flow cytometry. Expression of intracellular proteins was determined by Western blot. RESULTS: HSP90 is overexpressed in primary and cultured Hodgkin's lymphoma cells. Inhibition of HSP90 function by 17-AAG showed a time- and dose-dependent growth inhibition of Hodgkin's lymphoma cell lines. 17-AAG induced cell cycle arrest and apoptosis, which were associated with a decrease in cyclin-dependent kinase (CDK) 4, CDK 6, and polo-like kinase 1 (PLK1), and induced apoptosis by caspase-dependent and caspase-independent mechanisms. Furthermore, 17-AAG depleted cellular contents of Akt, decreased extracellular signal-regulated kinase (ERK) phosphorylation, and reduced cellular FLICE-like inhibitory protein levels (FLIP), and thus enhanced the cytotoxic effect of doxorubicin and agonistic anti-tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) death receptor antibodies. CONCLUSION: Inhibition of HSP90 function induces cell death and enhances the activity of chemotherapy and anti-tumor necrosis factor-related apoptosis-inducing ligand death receptor antibodies, suggesting that targeting HSP90 function might be of therapeutic value in Hodgkin's lymphoma.


Asunto(s)
Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Enfermedad de Hodgkin/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Rifabutina/análogos & derivados , Proteínas Adaptadoras Transductoras de Señales , Proteínas Reguladoras de la Apoptosis/metabolismo , Benzoquinonas , Western Blotting , Caspasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Regulación hacia Abajo , Doxorrubicina/farmacología , Citometría de Flujo , Proteínas HSP90 de Choque Térmico/metabolismo , Enfermedad de Hodgkin/metabolismo , Humanos , Lactamas Macrocíclicas , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Glicoproteínas de Membrana/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Rifabutina/farmacología , Transducción de Señal , Ligando Inductor de Apoptosis Relacionado con TNF , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/metabolismo , Quinasa Tipo Polo 1
15.
Exp Hematol ; 34(12): 1670-9, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17157164

RESUMEN

OBJECTIVE: Heat shock protein 90 (HSP90) chaperones and maintains the molecular integrity of a variety of signal transduction proteins, including the nucleophosmin-anaplastic lymphoma kinase (NPM-ALK) oncogenic protein, a genetic abnormality that is frequently observed in anaplastic large cell lymphoma (ALCL) cells. Here we demonstrate that HSP90 is overexpressed in primary and cultured ALK-positive and ALK-negative ALCL cells, and we evaluate the potential role of the small molecule inhibitor of HSP90, 17-allylamino-17-demethoxygeldanamycin (17-AAG) in treating ALCL. METHODS: The antiproliferative effect of 17-AAG-cultured cells was determined by MTS assay. Apoptosis and cell-cycle arrest were determined by Annexin-V/propidium iodide and propidium iodide staining, respectively, and fluorescein-activated cell sorting analysis. Expression of HSP90 was evaluated by immunohistochemistry, and molecular changes were determined by Western blot. RESULTS: Treatment of cultured ALCL cells with 17-AAG induced cell-cycle arrest and apoptosis, irrespective of ALK expression. At the molecular level, 17-AAG induced degradation of ALK and Akt proteins, dephosphorylated extracellular signal-regulated kinase, and degraded the cell-cycle regulatory protein cyclin D1 and its cyclin-dependent kinases, CDK4 and CDK6, but had a differential effect on p27 and p53 proteins. Inhibition of extracellular signal-regulated kinase phosphorylation by the mitogen activated protein kinase inhibitor U0126 induced cell death in all ALCL cell lines, and sublethal concentration 17-AAG showed synergistic antiproliferative effects when combined with U0126 or doxorubicin. CONCLUSION: Our data demonstrate that targeting HSP90 function by 17-AAG may offer a novel therapeutic strategy for ALCL, either as single-agent activity or by combining 17-AAG with conventional or targeted therapeutic schemes.


Asunto(s)
Benzoquinonas/farmacología , Butadienos/farmacología , Doxorrubicina/farmacología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Lactamas Macrocíclicas/farmacología , Linfoma Anaplásico de Células Grandes/metabolismo , Nitrilos/farmacología , Proteínas Tirosina Quinasas/biosíntesis , Quinasa de Linfoma Anaplásico , Apoptosis/efectos de los fármacos , Caspasa 3/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina D , Quinasa 4 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 6 Dependiente de la Ciclina/metabolismo , Ciclinas/efectos de los fármacos , Ciclinas/metabolismo , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Sinergismo Farmacológico , Quinasas MAP Reguladas por Señal Extracelular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fase G1/efectos de los fármacos , Proteínas HSP90 de Choque Térmico/biosíntesis , Humanos , Proteína Quinasa 1 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Proteínas Tirosina Quinasas/efectos de los fármacos , Proteínas Tirosina Quinasas Receptoras , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Factores de Tiempo
16.
Medicine (Baltimore) ; 96(49): e8599, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29245220

RESUMEN

RATIONALE: Heterotaxy with polysplenia is an extremely rare congenital condition resulting from abnormal arrangement of organs in the abdominal and thoracic cavities during embryologic development. When a malignancy such as pancreatic cancer develops under these conditions, surgical resection becomes particularly complex. This case report demonstrates successful pancreatic cancer resection despite the patient's complicated anatomy. PATIENT CONCERNS: An 82-year-old female presented to our institution with complaints of mild right upper quadrant pain radiating to the mid-epigastric region. DIAGNOSES: Physical examination revealed jaundice with scleral icterus consistent with obstructive jaundice. Radiographic imaging revealed hepatic duct dilation with several anatomic anomalies including small bowel location in the right upper abdomen, cecum, and appendix in the left lower quadrant, reversed superior mesenteric artery and superior mesenteric vein positions, and right-sided duodenal-jejunal flexture as well as an entirely right-sided pancreas, and left lower pelvis with ≥6 separate splenules. These findings resulted in a diagnosis of heterotaxy syndrome with polysplenia. INTERVENTIONS: Careful preoperative planning and total pancreatectomy was performed without complication. OUTCOMES: The patient recovered well. Pathologic examination of the pancreatic mass revealed moderately/poorly differentiated invasive pancreatic duct adenocarcinoma. The patient remains alive and well without signs of recurrent disease at the 2-year follow-up. LESSONS: Given the wide range of anatomical variants observed in patients with heterotaxy syndrome, a thorough radiologic assessment is necessary before engaging in any surgical procedure. In our case, preoperative identification of the various anatomic anomalies, such as the short and vertically oriented pancreas, the porta hepatis position anterior to the duodenum, the nonrotation of the intestines and the anomalous origin of the right hepatic artery allowed us to perform a safe and uncomplicated total pancreatectomy.


Asunto(s)
Síndrome de Heterotaxia/complicaciones , Vólvulo Intestinal/complicaciones , Pancreatectomía/métodos , Neoplasias Pancreáticas/cirugía , Bazo/anomalías , Anciano de 80 o más Años , Femenino , Humanos , Páncreas/cirugía , Neoplasias Pancreáticas/complicaciones
17.
Eur J Cancer ; 42(4): 542-7, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16426839

RESUMEN

The tumour necrosis factor (TNF) related apoptosis-inducing ligand (TRAIL/Apo2L) is a member of the TNF superfamily that preferentially induces apoptosis in cancer cells, while sparing normal cells. TRAIL induces apoptosis by interacting with its receptors TRAIL-R1 and TRAIL-R2. Recently, new humanized agonistic anti-TRAIL-R1 and anti-TRAIL-R2 antibodies have been developed, and are undergoing phase I/II clinical trails. Esophageal adenocarcinoma (EA) is associated with significantly poor outcome and is rapidly increasing in incidence in the United States and Western Europe, with virtually no effective non-surgical treatment. The aim of this study was to determine whether human EA tissue express TRAIL-R1 and/or TRAIL-R2, and whether EA cell lines Bic-1 and Seg-1 expresses functional TRAIL-R1 and/or TRAIL-R2. The expression of TRAIL-R1 and TRAIL-R2 was determined in sections from 18 human EA by immunohistochemistry (IHC). Sixteen (89%) of the EA expressed TRAIL-R1 and 17 (94%) expressed TRAIL-R2. Both cell lines were found to express TRAIL-R1 and TRAIL-R2 by western blot analysis, IHC, and flow cytometry. The fully human agonistic TRAIL-R1 (HGS-ETR1) and TRAIL-R2 (HGS-ETR2) antibodies induced apoptosis in Bic-1 and Seg-1 cells in a time and dose dependent manner. Our results show that the vast majority of primary human EA express TRAIL-R1 and TRAIL-R2 and that EA cells lines express functional TRAIL-R1 and TRAIL-R2. Targeting of these receptors by agonist monoclonal antibodies may be of therapeutic value in patients with EA.


Asunto(s)
Adenocarcinoma/metabolismo , Neoplasias Esofágicas/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Adenocarcinoma/patología , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos/farmacología , Apoptosis/inmunología , Línea Celular Tumoral , Neoplasias Esofágicas/patología , Citometría de Flujo , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF , Receptores del Factor de Necrosis Tumoral/inmunología
18.
Clin Cancer Res ; 10(9): 3207-15, 2004 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15131062

RESUMEN

PURPOSE: The malignant Hodgkin and Reed-Sternberg cells of Hodgkin disease (HD) are known to constitutively express high levels of activated nuclear factor kappaB (NF-kappaB), which plays an important role in their survival. The proteasome inhibitor PS-341 has been recently shown to modulate tumor cell proliferation and survival by inhibiting NF-kappaB and modulating critical cellular regulatory proteins, but its activity in cells carrying IkappaBalpha gene mutations has not been reported previously. EXPERIMENTAL DESIGN: The activity of PS-341 in four well-characterized, HD-derived cell lines. Cell proliferation and apoptosis were determined by the 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxy-phenyl)-2-(4-sulfonyl)-2H-tetrazolium (MTS) and Annexin-V binding methods, respectively. Cell cycle analysis was determined by flow cytometry. Intracellular protein levels were determined by Western blot. RESULTS: PS-341 demonstrated a strong antiproliferative activity, which was irrespective of the status of mutations in IkappaBalpha and even the presence of CD30, CD40, or RANK receptor activation. This effect was attributable to the induction of apoptosis and cell cycle arrest at the G(2)-M phase. PS-341 not only inhibited nuclear localization of NF-kappaB but also activated the caspase cascade, increased p21 and Bax levels, and decreased Bcl-2 levels. Furthermore, PS-341 enhanced the effect of gemcitabine chemotherapy and potentiated the effect of tumor necrosis factor-related apoptosis-inducing ligand/APO2L and two agonistic antibodies to tumor necrosis factor-related apoptosis-inducing ligand death receptors R1 and R2. CONCLUSIONS: The in vitro activity of PS-341 against HD-derived cell lines suggests that PS-341 may have a therapeutic value for the treatment of HD.


Asunto(s)
Apoptosis/efectos de los fármacos , Ácidos Borónicos/farmacología , Ciclo Celular/efectos de los fármacos , Inhibidores de Proteasas/farmacología , Pirazinas/farmacología , Anticuerpos Monoclonales/farmacología , Proteínas Reguladoras de la Apoptosis , Bortezomib , Antígenos CD40/metabolismo , Proteínas Portadoras/metabolismo , Caspasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , División Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cisteína Endopeptidasas , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Activación Enzimática/efectos de los fármacos , Enfermedad de Hodgkin/genética , Enfermedad de Hodgkin/metabolismo , Enfermedad de Hodgkin/patología , Humanos , Proteínas I-kappa B/genética , Antígeno Ki-1/metabolismo , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/farmacología , Complejos Multienzimáticos/antagonistas & inhibidores , Mutación , Inhibidor NF-kappaB alfa , FN-kappa B/metabolismo , Complejo de la Endopetidasa Proteasomal , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF , Receptores del Factor de Necrosis Tumoral/inmunología , Ligando Inductor de Apoptosis Relacionado con TNF , Factores de Tiempo , Factor de Transcripción ReIA , Factor de Necrosis Tumoral alfa/farmacología
20.
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA